Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Cell Sci ; 131(13)2018 07 06.
Article in English | MEDLINE | ID: mdl-29898922

ABSTRACT

Huntington's disease (HD) is a rare autosomal dominant neurodegenerative disorder caused by a cytosine-adenine-guanine (CAG) trinucleotide repeat (TNR) expansion within the HTT gene. The mechanisms underlying HD-associated cellular dysfunction in pluripotency and neurodevelopment are poorly understood. We had previously identified downregulation of selected DNA repair genes in HD fibroblasts relative to wild-type fibroblasts, as a result of promoter hypermethylation. Here, we tested the hypothesis that hypomethylation during cellular reprogramming to the induced pluripotent stem cell (iPSC) state leads to upregulation of DNA repair genes and stabilization of TNRs in HD cells. We sought to determine how the HD TNR region is affected by global epigenetic changes through cellular reprogramming and early neurodifferentiation. We find that early stage HD-affected neural stem cells (HD-NSCs) contain increased levels of global 5-hydroxymethylation (5-hmC) and normalized DNA repair gene expression. We confirm TNR stability is induced in iPSCs, and maintained in HD-NSCs. We also identify that upregulation of 5-hmC increases ten-eleven translocation 1 and 2 (TET1/2) protein levels, and show their knockdown leads to a corresponding decrease in the expression of select DNA repair genes. We further confirm decreased expression of TET1/2-regulating miR-29 family members in HD-NSCs. Our findings demonstrate that mechanisms associated with pluripotency induction lead to a recovery in the expression of select DNA repair gene and stabilize pathogenic TNRs in HD.


Subject(s)
DNA Repair , Epigenesis, Genetic , Huntington Disease/genetics , Induced Pluripotent Stem Cells/metabolism , Trinucleotide Repeat Expansion , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Neural Stem Cells/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
2.
Breast Cancer Res ; 20(1): 122, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30305139

ABSTRACT

BACKGROUND: Standard three-dimensional (3D) in vitro culture techniques, such as those used for mammary epithelial cells, rely on random distribution of cells within hydrogels. Although these systems offer advantages over traditional 2D models, limitations persist owing to the lack of control over cellular placement within the hydrogel. This results in experimental inconsistencies and random organoid morphology. Robust, high-throughput experimentation requires greater standardization of 3D epithelial culture techniques. METHODS: Here, we detail the use of a 3D bioprinting platform as an investigative tool to control the 3D formation of organoids through the "self-assembly" of human mammary epithelial cells. Experimental bioprinting procedures were optimized to enable the formation of controlled arrays of individual mammary organoids. We define the distance and cell number parameters necessary to print individual organoids that do not interact between print locations as well as those required to generate large contiguous organoids connected through multiple print locations. RESULTS: We demonstrate that as few as 10 cells can be used to form 3D mammary structures in a single print and that prints up to 500 µm apart can fuse to form single large structures. Using these fusion parameters, we demonstrate that both linear and non-linear (contiguous circles) can be generated with sizes of 3 mm in length/diameter. We confirm that cells from individual prints interact to form structures with a contiguous lumen. Finally, we demonstrate that organoids can be printed into human collagen hydrogels, allowing for all-human 3D culture systems. CONCLUSIONS: Our platform is adaptable to different culturing protocols and is superior to traditional random 3D culture techniques in efficiency, reproducibility, and scalability. Importantly, owing to the low-cost accessibility and computer numerical control-driven platform of our 3D bioprinter, we have the ability to disseminate our experiments with absolute precision to interested laboratories.


Subject(s)
Bioprinting/methods , Cell Culture Techniques/methods , Epithelial Cells/cytology , Mammary Glands, Human/cytology , Cell Line , Female , Humans , Hydrogels , Organoids/cytology , Organoids/growth & development , Reproducibility of Results
3.
Breast Cancer Res ; 20(1): 136, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30454070

ABSTRACT

Following publication of the original article [1], the authors reported a typesetting error in the spelling of the second author's name.

4.
Am J Pathol ; 186(7): 1967-1976, 2016 07.
Article in English | MEDLINE | ID: mdl-27182645

ABSTRACT

Huntington disease (HD) is an autosomal dominantly inherited disease that exhibits genetic anticipation of affected progeny due to expansions of a trinucleotide repeat (TNR) region within the HTT gene. DNA repair machinery is a known effector of TNR instability; however, the specific defects in HD cells that lead to TNR expansion are unknown. We hypothesized that HD cells would be deficient in DNA repair gene expression. To test this hypothesis, we analyzed expression of select DNA repair genes involved in mismatch/loop-out repair (APEX1, BRCA1, RPA1, and RPA3) in patient-derived HD cells and found each was consistently down-regulated relative to wild-type samples taken from unaffected individuals in the same family. Rescue of DNA repair gene expression by 5-azacytidine treatment identified DNA methylation as a mediator of DNA repair gene expression deficiency. Bisulfite sequencing confirmed hypermethylation of the APEX1 promoter region in HD cells relative to control, as well as 5-azacytidine-induced hypomethylation. 5-Azacytidine treatments also resulted in stabilization of TNR expansion within the mutant HTT allele during long-term culture of HD cells. Our findings indicate that DNA methylation leads to DNA repair down-regulation and TNR instability in mitotically active HD cells and offer a proof of principle that epigenetic interventions can curb TNR expansions.


Subject(s)
DNA Methylation/genetics , DNA Repair/genetics , Huntington Disease/genetics , Trinucleotide Repeat Expansion/genetics , Cells, Cultured , Down-Regulation , Fluorescent Antibody Technique , Humans , Real-Time Polymerase Chain Reaction
5.
J Cell Sci ; 127(Pt 1): 27-32, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24190884

ABSTRACT

We have previously shown that non-mammary and tumorigenic cells can respond to the signals of the mammary niche and alter their cell fate to that of mammary epithelial progenitor cells. Here we tested the hypothesis that paracrine signals from mammary epithelial cells expressing progesterone receptor (PR) are dispensable for redirection of testicular cells, and that re-directed wild-type testicular-derived mammary cells can rescue lobulogenesis of PR-null mammary epithelium by paracrine signaling during pregnancy. We injected PR-null epithelial cells mixed with testicular cells from wild-type adult male mice into cleared fat-pads of recipient mice. The testicular cells were redirected in vivo to mammary epithelial cell fate during regeneration of the mammary epithelium, and persisted in second-generation outgrowths. In the process, the redirected testicular cells rescued the developmentally deficient PR-null cells, signaling them through the paracrine factor RANKL to produce alveolar secretory structures during pregnancy. This is the first demonstration that paracrine signaling required for alveolar development is not required for cellular reprogramming in the mammary gland, and that reprogrammed testicular cells can provide paracrine signals to the surrounding mammary epithelium.


Subject(s)
Cellular Reprogramming/genetics , Epithelial Cells/cytology , Mammary Glands, Animal/cytology , Paracrine Communication/genetics , Receptors, Progesterone/genetics , Seminiferous Tubules/cytology , Adipose Tissue , Animals , Cell Differentiation , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Female , Gene Expression , Injections , Male , Mammary Glands, Animal/metabolism , Mice , Pregnancy , Progesterone/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Receptors, Progesterone/deficiency , Seminiferous Tubules/metabolism , Seminiferous Tubules/transplantation , Signal Transduction
6.
Semin Cell Dev Biol ; 23(5): 591-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22430755

ABSTRACT

The capacity of any portion of the murine mammary gland to produce a complete functional mammary outgrowth upon transplantation to an epithelium-divested fat pad is unaffected by the age or reproductive history of the donor. Likewise, through serial transplantations, no loss of potency is detected when compared to similar transplantations of the youngest mammary tissue tested. This demonstrates that stem cell activity is maintained intact throughout the lifetime of the animal despite aging and the repeated expansion and depletion of the mammary epithelium through multiple rounds of pregnancy, lactation and involution. These facts support the contention that mammary stem cells reside in protected tissue locales (niches), where their reproductive potency remains essentially unchanged through life. Disruption of the tissue, to produce dispersed cells results in the desecration of the protection afforded by the "niche" and leads to a reduced capacity of dispersed epithelial cells (in terms of the number transplanted) to recapitulate complete functional mammary structures. Our studies demonstrate that during the reformation of mammary stem cell niches by dispersed epithelial cells in the context of the intact epithelium-free mammary stroma, non-mammary cells, including mouse and human cancer cells, may be sequestered and reprogrammed to perform mammary epithelial cell functions including those ascribed to mammary stem/progenitor cells.


Subject(s)
Cellular Reprogramming , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Neoplasms/metabolism , Animals , Humans , Mammary Glands, Animal/growth & development , Mice , Mice, Transgenic , Neoplasms/pathology , Stem Cells/cytology , Stem Cells/metabolism
7.
Breast Cancer Res ; 16(1): 302, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-25927296

ABSTRACT

Extracellular matrix proteins from embryonic mesenchyme have a normalizing effect on cancer cells in vitro and slow tumor growth in vivo. This concept is suggestive of a new method for controlling the growth and spread of existing cancer cells in situ and indicates the possibility that extracellular proteins and/or embryonic mesenchymal fibroblasts may represent a fertile subject for study of new anti-cancer treatments.


Subject(s)
Biglycan/chemistry , Breast Neoplasms/pathology , Cell Culture Techniques , Extracellular Matrix/metabolism , Mammary Glands, Animal/embryology , Mesoderm/pathology , Animals , Female , Humans
8.
Mol Cancer ; 12: 79, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23866257

ABSTRACT

BACKGROUND: The canonical milk-transmitted mouse mammary tumor virus (MMTV) of C3H mice (C3H-MMTV) rapidly induces tumors in 90% of infected animals by 8 months of age. Pro-viral insertions of C3H-MMTV into genomic DNA results in the overexpression of common core insertion site (CIS) genes, including Wnt1/10b, Rspo2, and Fgf3. Conversely, infection by either the endogenous Mtv-1 virus (in C3Hf) or the exogenous nodule-inducing virus (NIV) (in Balb/c NIV) induces premalignant mammary lesions and tumors with reduced incidence and longer latency than C3H-MMTV. Here, we asked whether Mtv-1/NIV affected the expression of core CIS genes. FINDINGS: We confirmed the presence of active virus in Mtv-1/NIV infected tissues and using quantitative reverse transcription PCR (qRT-PCR) found that Mtv-1/NIV induced neoplasms (tumors and hyperplasia) commonly expressed the core CIS genes Wnt1, Wnt10b, Rspo2, Fgf3. CONCLUSIONS: These results underscore the importance of core CIS gene expression in the early events leading to MMTV-induced mammary tumor initiation regardless of the viral variant.


Subject(s)
Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/physiology , Animals , Female , Hyperplasia , Mammary Neoplasms, Experimental/virology , Mice , Mice, Inbred BALB C
9.
Front Cell Dev Biol ; 11: 1258993, 2023.
Article in English | MEDLINE | ID: mdl-37928905

ABSTRACT

We have previously shown that human and murine breast extracellular matrix (ECM) can significantly impact cellular behavior, including stem cell fate determination. It has been established that tissue-specific extracellular matrix from the central nervous system has the capacity to support neuronal survival. However, the characterization of its influence on stem cell differentiation and its adaptation to robust 3D culture models is underdeveloped. To address these issues, we combined our 3D bioprinter with hydrogels containing porcine brain extracellular matrix (BMX) to test the influence of the extracellular matrix on stem cell differentiation. Our 3D bioprinting system generated reproducible 3D neural structures derived from mouse embryonic stem cells (mESCs). We demonstrate that the addition of BMX preferentially influences 3D bioprinted mESCs towards neural lineages compared to standard basement membrane (Geltrex/Matrigel) hydrogels alone. Furthermore, we demonstrate that we can transplant these 3D bioprinted neural cellular structures into a mouse's cleared mammary fat pad, where they continue to grow into larger neural outgrowths. Finally, we demonstrate that direct injection of human induced pluripotent stem cells (hiPSCS) and neural stem cells (NSCs) suspended in pure BMX formed neural structures in vivo. Combined, these findings describe a unique system for studying brain ECM/stem cell interactions and demonstrate that BMX can direct pluripotent stem cells to differentiate down a neural cellular lineage without any additional specific differentiation stimuli.

10.
Gene Expr ; 15(3): 133-40, 2011.
Article in English | MEDLINE | ID: mdl-22268295

ABSTRACT

Both mouse and human mammary glands contain stem/progenitor functional hierarchies that are maintained through the entire life span of the animal. Cells with such functional capacities are potential candidates for tumorigenesis as they are long lived, multipotent, and self-renewing. Using the mouse as a model, this review will discuss what is known about the mammary stem/progenitor hierarchy, the evidence that particular progenitor functions are susceptible to tumorigenic stimuli, how these findings in mice are relevant to the disease in humans, and the role of the local microenvironment in controlling tumorigenesis.


Subject(s)
Breast Neoplasms/pathology , Epithelial Cells/physiology , Mammary Glands, Human/cytology , Mammary Glands, Human/pathology , Stem Cells/physiology , Animals , Breast Neoplasms/physiopathology , Cell Transformation, Neoplastic , Disease Models, Animal , Epithelial Cells/cytology , Female , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/pathology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Stem Cell Niche , Stem Cells/cytology , Tumor Microenvironment
11.
Mol Cancer Ther ; 7(9): 2828-36, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18790763

ABSTRACT

Inhibitors of the enzyme 17alpha-hydroxylase/17,20 lyase are a new class of anti-prostate cancer agents currently undergoing preclinical and clinical development. We have previously reported the superior anticancer activity of our novel 17alpha-hydroxylase/17,20 lyase inhibitor, VN/124-1, against androgen-dependent cancer models. Here, we examined the effect of VN/124-1 on the growth of the androgen-independent cell lines PC-3 and DU-145 and found that the compound inhibits their growth in a dose-dependent manner in vitro (GI50, 7.82 micromol/L and 7.55 micromol/L, respectively). We explored the mechanism of action of VN/124-1 in PC-3 cells through microarray analysis and found that VN/124-1 up-regulated genes involved in stress response and protein metabolism, as well as down-regulated genes involved in cell cycle progression. Follow-up real-time PCR and Western blot analyses revealed that VN/124-1 induces the endoplasmic reticulum stress response resulting in down-regulation of cyclin D1 protein expression and cyclin E2 mRNA. Cell cycle analysis confirmed G1-G0 phase arrest. Measurements of intracellular calcium levels ([Ca2+]i) showed that 20 micromol/L VN/124-1 caused a release of Ca2+ from endoplasmic reticulum stores resulting in a sustained increase in [Ca2+]i. Finally, cotreatment of PC-3 cells with 5, 10, and 20 micromol/L VN/124-1 with 10 nmol/L thapsigargin revealed a synergistic relationship between the compounds in inhibiting PC-3 cell growth. Taken together, these findings show VN/124-1 is endowed with multiple anticancer properties that may contribute to its utility as a prostate cancer therapeutic.


Subject(s)
Androstadienes/pharmacology , Benzimidazoles/pharmacology , Endoplasmic Reticulum/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Androgens/metabolism , Androstadienes/chemistry , Animals , Benzimidazoles/chemistry , Calcium/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/genetics , Down-Regulation/drug effects , Drug Synergism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Eukaryotic Initiation Factor-2/metabolism , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm , Male , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Prostatic Neoplasms/genetics , Resting Phase, Cell Cycle/drug effects , Thapsigargin/pharmacology , Up-Regulation/drug effects
12.
Mech Dev ; 159: 103565, 2019 10.
Article in English | MEDLINE | ID: mdl-31336167

ABSTRACT

Long-label retention has been used by many to prove Cairns' immortal strand hypothesis and to identify potential stem cells. Here, we describe two strategies using 5-ethynl-2'-deoxyuridine (EdU) to identify and understand the distribution of long-label-retaining mammary epithelial cells during formation of the mouse mammary ductal system. First, EdU was given upon two consecutive days per week during weeks 4 through 10 and analyzed for label retention at 13 weeks of age. Alternatively, EdU was given for 14 consecutive days beginning at 28 days of age and ending at 42 days of age. Analyses were conducted at >91 days of age (13 weeks). Many more LREC were detected following the second labeling method and their distribution among the subsequently developed ducts. This finding indicated that the early-labeled cells that retained their label were distributed into portions of the gland that developed after the ending of EdU treatment (i.e. 42->91 days). These observations may have important meaning with respect to the previously demonstrated retention of regenerative capacity throughout the mouse mammary gland despite age or reproductive history. These results suggest LREC may represent long-lived progenitor cells that are responsible for mammary gland homeostasis. Additionally, these cells may act as multipotent stem cells capable of mammary gland regeneration upon random fragment transplantation into epithelium-denuded mammary fat pads.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Organogenesis , Animals , Female , G2 Phase , Mice, Inbred BALB C , Mice, Nude , Staining and Labeling
13.
Sci Rep ; 9(1): 7466, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097753

ABSTRACT

The normal mammary microenvironment can suppress tumorigenesis and redirect cancer cells to adopt a normal mammary epithelial cell fate in vivo. Understanding of this phenomenon offers great promise for novel treatment and detection strategies in cancer, but current model systems make mechanistic insights into the process difficult. We have recently described a low-cost bioprinting platform designed to be accessible for basic cell biology laboratories. Here we report the use of this system for the study of tumorigenesis and microenvironmental redirection of breast cancer cells. We show our bioprinter significantly increases tumoroid formation in 3D collagen gels and allows for precise generation of tumoroid arrays. We also demonstrate that we can mimic published in vivo findings by co-printing cancer cells along with normal mammary epithelial cells to generate chimeric organoids. These chimeric organoids contain cancer cells that take part in normal luminal formation. Furthermore, we show for the first time that cancer cells within chimeric structures have a significant increase in 5-hydroxymethylcytosine levels as compared to bioprinted tumoroids. These results demonstrate the capacity of our 3D bioprinting platform to study tumorigenesis and microenvironmental control of breast cancer and highlight a novel mechanistic insight into the process of microenvironmental control of cancer.


Subject(s)
Breast Neoplasms/pathology , Organoids/pathology , Printing, Three-Dimensional , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Biotechnology/methods , Carcinogenesis/pathology , Cell Line , Cells, Cultured , Female , Humans , MCF-7 Cells , Mammary Glands, Human/cytology , Mammary Glands, Human/pathology , Organoids/metabolism , Tumor Microenvironment
14.
Acta Biomater ; 95: 201-213, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31233891

ABSTRACT

The extracellular matrix (ECM) of tissues is an important mediator of cell function. Moreover, understanding cellular dynamics within their specific tissue context is also important for developmental biology, cancer research, and regenerative medicine. However, robust in vitro models that incorporate tissue-specific microenvironments are lacking. Here we describe a novel mammary-specific culture protocol that combines a self-gelling hydrogel comprised solely of ECM from decellularized rat or human breast tissue with the use of our previously described 3D bioprinting platform. We initially demonstrate that undigested and decellularized mammary tissue can support mammary epithelial and tumor cell growth. We then describe a methodology for generating mammary ECM extracts that can spontaneously gel to form hydrogels. These ECM hydrogels retain unique structural and signaling profiles that elicit differential responses when normal mammary and breast cancer cells are cultured within them. Using our bioprinter, we establish that we can generate large organoids/tumoroids in the all mammary-derived hydrogel. These findings demonstrate that our system allows for growth of organoids/tumoroids in a tissue-specific matrix with unique properties, thus providing a suitable platform for ECM and epithelial/cancer cell studies. STATEMENT OF SIGNIFICANCE: Factors within extracellular matrices (ECMs) are specific to their tissue of origin. It has been shown that tissue specific factors within the mammary gland's ECM have pronounced effects on cellular differentiation and cancer behavior. Understanding the role of the ECM in controlling cell fate has major implications for developmental biology, tissue engineering, and cancer therapy. However, in vitro models to study cellular interactions with tissue specific ECM are lacking. Here we describe the generation of 3D hydrogels consisting solely of human or mouse mammary ECM. We demonstrate that these novel 3D culture substrates can sustain large 3D bioprinted organoid and tumoroid formation. This is the first demonstration of an all mammary ECM culture system capable of sustaining large structural growths.


Subject(s)
Bioprinting , Breast Neoplasms/pathology , Extracellular Matrix/chemistry , Hydrogels/pharmacology , Mammary Glands, Human/pathology , Organoids/metabolism , Printing, Three-Dimensional , Animals , Cell Line, Tumor , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Keratin-5/metabolism , Ki-67 Antigen/metabolism , Rats , Signal Transduction
15.
Bioorg Med Chem ; 16(7): 3519-29, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18316193

ABSTRACT

The search for novel androgen receptor (AR) down-regulating agents by catalyst HipHop pharmacophore modeling led to the discovery of some lead molecules. Unexpectedly, the effect of these leads on human prostate cancer LNCaP cell viability did not correlate with the ability of the compounds to cause down-regulation of AR protein expression. Through rational synthetic optimization of the lead compound (BTB01434), we have discovered a series of novel substituted diaryl molecules as potent anti-prostate cancer agents. Some compounds (1-6) were shown to be extremely potent inhibitors of LNCaP cell viability with GI(50) values in the nanomolar range (1.45-83 nM). The most potent compound (4-methylphenyl)[(4-methylphenyl)sulfonyl]amine (5) with a GI(50) value of 1.45 nM is 27,000 times more potent than our lead compound BTB01434 (GI(50)=39.8 microM). In addition, some of the compounds exhibited modest anti-androgenic activities and one was also a potent inhibitor (GI(50)=850 nM) of PC-3 (AR-null) cell growth. A clear structure-activity relationship (SAR) has been established for activity against LNCaP cells, where potent molecules possess two substituted/unsubstituted aromatic rings connected through a sulfonamide linker. These novel compounds are strong candidates for development for the treatment of hormone-sensitive and importantly hormone-refractory prostate cancers in humans.


Subject(s)
Androgen Receptor Antagonists , Antineoplastic Agents, Hormonal/chemical synthesis , Antineoplastic Agents, Hormonal/pharmacology , Down-Regulation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Amines/chemical synthesis , Amines/chemistry , Androgens/metabolism , Antineoplastic Agents, Hormonal/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Humans , Imines/chemical synthesis , Imines/chemistry , Male , Models, Molecular , Molecular Structure , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Structure-Activity Relationship , Transcription, Genetic/genetics
16.
Int Urol Nephrol ; 40(3): 749-55, 2008.
Article in English | MEDLINE | ID: mdl-18438718

ABSTRACT

BACKGROUND: The effect of continuing or discontinuing chronic angiotensin-converting enzyme inhibitor (ACEI) or angiotensin receptor blocker (ARB) therapy prior to coronary angiography on the incidence of contrast-induced nephropathy (CIN) is not clear. We undertook a randomized trial to evaluate the effect of withdrawing ACEIs or ARBs 24 h prior to coronary angiography on the incidence of CIN associated with coronary angiography. METHODS: A total of 220 patients with chronic kidney disease (CKD) stages 3-4 (glomerular filtration rate 15-60 ml/min/1.73 m2) on ACEI or ARB therapy were randomized before angiography to either ACEI/ARB continuation group or discontinuation group. A third group of patients with CKD stages 3-4 but not on angiotensin blockade therapy were also followed. The primary outcome measure was the incidence of CIN defined by a rise in serum creatinine by 25% or 0.5 mg/dl (44 micromol/l) from baseline. RESULTS: There was no statistically significant difference in the incidence of CIN between the three groups (P=0.66). The incidences were 6.2%, 3.7%, and 6.3% for the continuation, discontinuation, and angiotensin blockade naïve group, respectively. There was also no significant difference found between the groups in mean serum creatinine and glomerular filtration rate values at baseline and post contrast administration. CONCLUSION: Withholding ACEIs and ARBs 24 h before coronary angiography does not appear to influence the incidence of CIN in stable patients with CKD stages 3-4.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Contrast Media/adverse effects , Coronary Angiography , Kidney Diseases/chemically induced , Kidney Failure, Chronic/complications , Aged , Analysis of Variance , Creatinine/blood , Female , Humans , Incidence , Male
17.
Pathology ; 50(5): 524-529, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29891189

ABSTRACT

Interest into the cellular biology of human tonsillar crypts has grown in recent years because it is now known to be the site of origin of most human papilloma virus (HPV) induced oropharyngeal squamous cell carcinomas (OPSCC). Despite the interest, still relatively little is known regarding the cellular hierarchy and dynamics of this anatomical subsite. Here we evaluate normal tonsillar crypts for expression of putative stem cell markers. We found that ALDH1A1 was uniquely expressed in a subset of suprabasal tonsillar crypt epithelium. This cell population was unique from NGFR expressing cells, which were previously identified to have stem/progenitor activity in vitro. In vivo mitochondrial lineage tracing was consistent with a basal to luminal progression of cellular development. This provides support for NGFR cells as the resident stem/progenitor cells in tonsillar crypts, and suggests that the ALDH1A1 cells are not stem/progenitor cells, but merely a unique component of the crypt cellular microenvironment. Analysis of tumours found that both NGFR and ALDH1A1 are lost in HPV+ and HPV- tumours, while LGR5 expression is induced in the same tumours. These results identify a unique component of the tonsillar crypt epithelium-ALDH1A1 cells-and support a cellular model where NGFR+ cells are the long-lived progenitor cells within tonsillar crypts. They also provide evidence that NGFR and ALDH1A1+ cells are lost during tumourigenesis.


Subject(s)
Aldehyde Dehydrogenase/genetics , Carcinoma, Squamous Cell/genetics , Cell Transformation, Neoplastic/genetics , Head and Neck Neoplasms/genetics , Palatine Tonsil/metabolism , Aldehyde Dehydrogenase 1 Family , Carcinoma, Squamous Cell/pathology , Cell Differentiation/physiology , Cell Transformation, Neoplastic/pathology , Head and Neck Neoplasms/pathology , Humans , Nerve Tissue Proteins/genetics , Papillomavirus Infections/pathology , Receptors, Nerve Growth Factor/genetics , Retinal Dehydrogenase , Squamous Cell Carcinoma of Head and Neck
18.
J Neural Eng ; 15(5): 056021, 2018 10.
Article in English | MEDLINE | ID: mdl-29848804

ABSTRACT

OBJECTIVE: Picosecond pulse electric fields (psPEF) have the potential to elicit functional changes in mammalian cells in a non-contact manner. Such electro-manipulation of pluripotent and multipotent cells could be a tool in both neural interface and tissue engineering. Here, we describe the potential of psPEF in directing neural stem cells (NSCs) gene expression, metabolism, and proliferation. As a comparison mesenchymal stem cells (MSCs) were also tested. APPROACH: A psPEF electrode was anchored on a customized commercially available 3D printer, which allowed us to deliver pulses with high spatial precision and systematically control the electrode position in three-axes. When the electrodes are continuously energized and their position is shifted by the 3D printer, large numbers of cells on a surface can be exposed to a uniform psPEF. With two electric field strengths (20 and 40 kV cm-1), cell responses, including cell viability, proliferation, and gene expression assays, were quantified and analyzed. MAIN RESULTS: Analysis revealed both NSCs and MSCs showed no significant cell death after treatments. Both cell types exhibited an increased metabolic reduction; however, the response rate for MSCs was sensitive to the change of electric field strength, but for NSCs, it appeared independent of electric field strength. The change in proliferation rate was cell-type specific. MSCs underwent no significant change in proliferation whereas NSCs exhibited an electric field dependent response with the higher electric field producing less proliferation. Further, NSCs showed an upregulation of glial fibrillary acidic protein (GFAP) after 24 h to 40 kV cm-1, which is characteristic of astrocyte specific differentiation. SIGNIFICANCE: Changes in cell metabolism, proliferation, and gene expression after picosecond pulsed electric field exposure are cell type specific.


Subject(s)
Cell Lineage/genetics , Cell Proliferation , Electromagnetic Fields , Gene Expression/genetics , Neural Stem Cells/physiology , Printing, Three-Dimensional , Astrocytes/metabolism , Cell Death , Electrodes , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Humans , Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Neurogenesis
19.
Biochim Biophys Acta ; 1760(11): 1646-56, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17049166

ABSTRACT

By selection of genetic suppressor elements (GSEs) conferring resistance to topoisomerase II inhibitors in Chinese hamster cells (DC-3F), we identified a gene encoding two proteins of 78 and 82 kDa which belong to the protein arginine methyltransferase (PRMT) family. Down-regulation of these enzymes (named PRMT7alpha and beta), either induced by an antisense GSE or as observed in the 9-OH-ellipticine (9-OH-E) resistant mutant DC-3F/9-OH-E, was responsible for cell resistance to various DNA damaging agents. Alternative splicing alterations in the 5'-terminal region and changes of the polyadenylation site of PRMT7 mRNAs were observed in these resistant mutant cells. PRMT7alpha and beta are isoforms of a highly conserved protein containing two copies of a module common to all PRMTs, comprising a Rossmann-fold domain and a beta-barrel domain. The C-terminal repeat appears to be degenerate and catalytically inactive. PRMT7alpha and beta form homo- and hetero-dimers but differ by their sub-cellular localization and in vitro recognize different substrates. PRMT7beta was only observed in Chinese hamster cells while mouse 10T1/2 fibroblasts only contain PRMT7alpha. Surprisingly, in human cells the anti-PRMT7 antibody essentially recognized an approximately 37 kDa peptide, which is not formed during extraction, and a faint band at 78 kDa. Analysis of in vitro and in vivo methylation patterns in cell lines under- or over-expressing PRMT7alpha and beta detected a discrete number of proteins which methylation and/or expression are under the control of these enzymes.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/genetics , Topoisomerase II Inhibitors , Animals , Cell Cycle , Cricetinae , Cricetulus , DNA Topoisomerases, Type II/metabolism , Dimerization , HeLa Cells , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Methylation , Methyltransferases/chemistry , Methyltransferases/genetics , Mice , Protein-Arginine N-Methyltransferases/metabolism , Species Specificity , Tumor Cells, Cultured
20.
J Biol Eng ; 11: 34, 2017.
Article in English | MEDLINE | ID: mdl-29177006

ABSTRACT

The accumulated evidence points to the microenvironment as the primary mediator of cellular fate determination. Comprised of parenchymal cells, stromal cells, structural extracellular matrix proteins, and signaling molecules, the microenvironment is a complex and synergistic edifice that varies tissue to tissue. Furthermore, it has become increasingly clear that the microenvironment plays crucial roles in the establishment and progression of diseases such as cardiovascular disease, neurodegeneration, cancer, and ageing. Here we review the historical perspectives on the microenvironment, and how it has directed current explorations in tissue engineering. By thoroughly understanding the role of the microenvironment, we can begin to correctly manipulate it to prevent and cure diseases through regenerative medicine techniques.

SELECTION OF CITATIONS
SEARCH DETAIL